Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Genome Res ; 33(6): 857-871, 2023 06.
Article in English | MEDLINE | ID: mdl-37217254

ABSTRACT

The Diversity Outbred (DO) mice and their inbred founders are widely used models of human disease. However, although the genetic diversity of these mice has been well documented, their epigenetic diversity has not. Epigenetic modifications, such as histone modifications and DNA methylation, are important regulators of gene expression and, as such, are a critical mechanistic link between genotype and phenotype. Therefore, creating a map of epigenetic modifications in the DO mice and their founders is an important step toward understanding mechanisms of gene regulation and the link to disease in this widely used resource. To this end, we performed a strain survey of epigenetic modifications in hepatocytes of the DO founders. We surveyed four histone modifications (H3K4me1, H3K4me3, H3K27me3, and H3K27ac), as well as DNA methylation. We used ChromHMM to identify 14 chromatin states, each of which represents a distinct combination of the four histone modifications. We found that the epigenetic landscape is highly variable across the DO founders and is associated with variation in gene expression across strains. We found that epigenetic state imputed into a population of DO mice recapitulated the association with gene expression seen in the founders, suggesting that both histone modifications and DNA methylation are highly heritable mechanisms of gene expression regulation. We illustrate how DO gene expression can be aligned with inbred epigenetic states to identify putative cis-regulatory regions. Finally, we provide a data resource that documents strain-specific variation in the chromatin state and DNA methylation in hepatocytes across nine widely used strains of laboratory mice.


Subject(s)
DNA Methylation , Histones , Humans , Mice , Animals , Histones/genetics , Histones/metabolism , Promoter Regions, Genetic , Chromatin/genetics , Epigenesis, Genetic , Histone Code , Mice, Inbred Strains , Gene Expression
2.
Nucleic Acids Res ; 51(11): 5364-5376, 2023 06 23.
Article in English | MEDLINE | ID: mdl-36951113

ABSTRACT

The human genome contains about 800 C2H2 zinc finger proteins (ZFPs), and most of them are composed of long arrays of zinc fingers. Standard ZFP recognition model asserts longer finger arrays should recognize longer DNA-binding sites. However, recent experimental efforts to identify in vivo ZFP binding sites contradict this assumption, with many exhibiting short motifs. Here we use ZFY, CTCF, ZIM3, and ZNF343 as examples to address three closely related questions: What are the reasons that impede current motif discovery methods? What are the functions of those seemingly unused fingers and how can we improve the motif discovery algorithms based on long ZFPs' biophysical properties? Using ZFY, we employed a variety of methods and find evidence for 'dependent recognition' where downstream fingers can recognize some previously undiscovered motifs only in the presence of an intact core site. For CTCF, high-throughput measurements revealed its upstream specificity profile depends on the strength of its core. Moreover, the binding strength of the upstream site modulates CTCF's sensitivity to different epigenetic modifications within the core, providing new insight into how the previously identified intellectual disability-causing and cancer-related mutant R567W disrupts upstream recognition and deregulates the epigenetic control by CTCF. Our results establish that, because of irregular motif structures, variable spacing and dependent recognition between sub-motifs, the specificities of long ZFPs are significantly underestimated, so we developed an algorithm, ModeMap, to infer the motifs and recognition models of ZIM3 and ZNF343, which facilitates high-confidence identification of specific binding sites, including repeats-derived elements. With revised concept, technique, and algorithm, we can discover the overlooked specificities and functions of those 'extra' fingers, and therefore decipher their broader roles in human biology and diseases.


Subject(s)
DNA , Transcription Factors , Zinc Fingers , Humans , Binding Sites , Transcription Factors/chemistry , Transcription Factors/metabolism , Algorithms , Nucleotide Motifs , Amino Acid Motifs , DNA/chemistry , DNA/metabolism
3.
Cell Mol Life Sci ; 80(4): 107, 2023 Mar 26.
Article in English | MEDLINE | ID: mdl-36967403

ABSTRACT

In mammals, meiotic recombination is initiated by the introduction of DNA double strand breaks (DSBs) into narrow segments of the genome, defined as hotspots, which is carried out by the SPO11/TOPOVIBL complex. A major player in the specification of hotspots is PRDM9, a histone methyltransferase that, following sequence-specific DNA binding, generates trimethylation on lysine 4 (H3K4me3) and lysine 36 (H3K36me3) of histone H3, thus defining the hotspots. PRDM9 activity is key to successful meiosis, since in its absence DSBs are redirected to functional sites and synapsis between homologous chromosomes fails. One protein factor recently implicated in guiding PRDM9 activity at hotspots is EWS, a member of the FET family of proteins that also includes TAF15 and FUS/TLS. Here, we demonstrate that FUS/TLS partially colocalizes with PRDM9 on the meiotic chromosome axes, marked by the synaptonemal complex component SYCP3, and physically interacts with PRDM9. Furthermore, we show that FUS/TLS also interacts with REC114, one of the axis-bound SPO11-auxiliary factors essential for DSB formation. This finding suggests that FUS/TLS is a component of the protein complex that promotes the initiation of meiotic recombination. Accordingly, we document that FUS/TLS coimmunoprecipitates with SPO11 in vitro and in vivo. The interaction occurs with both SPO11ß and SPO11α splice isoforms, which are believed to play distinct functions in the formation of DSBs in autosomes and male sex chromosomes, respectively. Finally, using chromatin immunoprecipitation experiments, we show that FUS/TLS is localized at H3K4me3-marked hotspots in autosomes and in the pseudo-autosomal region, the site of genetic exchange between the XY chromosomes.


Subject(s)
Lysine , RNA-Binding Protein FUS , Animals , Male , Lysine/genetics , RNA-Binding Protein FUS/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Homologous Recombination , DNA/metabolism , Meiosis/genetics , Mammals/metabolism
4.
Proc Natl Acad Sci U S A ; 119(32): e2123362119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35921433

ABSTRACT

The germinal center (GC) plays a central role in the generation of antigen-specific B cells and antibodies. Tight regulation of the GC is essential due to the inherent risks of tumorigenesis and autoimmunity posed by inappropriate GC B cell processes. Gammaherpesviruses such as Epstein-Barr virus (EBV) and murine gammaherpesvirus 68 (MHV68) utilize numerous armaments to drive infected naïve B cells, independent of antigen, through GC reactions to expand the latently infected B cell population and establish a stable latency reservoir. We previously demonstrated that the MHV68 microRNA (miRNA) mghv-miR-M1-7-5p represses host EWSR1 (Ewing sarcoma breakpoint region 1) to promote B cell infection. EWSR1 is a transcription and splicing regulator that is recognized for its involvement as a fusion protein in Ewing sarcoma. A function for EWSR1 in B cell responses has not been previously reported. Here, we demonstrate that 1) B cell-specific deletion of EWSR1 had no effect on generation of mature B cell subsets or basal immunoglobulin levels in naïve mice, 2) repression or ablation of EWSR1 in B cells promoted expansion of MHV68 latently infected GC B cells, and 3) B cell-specific deletion of EWSR1 during a normal immune response to nonviral antigen resulted in significantly elevated numbers of antigen-specific GC B cells, plasma cells, and circulating antibodies. Notably, EWSR1 deficiency did not affect the proliferation or survival of GC B cells but instead resulted in the generation of increased numbers of precursor GC B cells. Cumulatively, these findings demonstrate that EWSR1 is a negative regulator of B cell responses.


Subject(s)
B-Lymphocytes , Gammaherpesvirinae , Germinal Center , Herpesviridae Infections , MicroRNAs , RNA-Binding Protein EWS , Tumor Virus Infections , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , Gammaherpesvirinae/genetics , Gammaherpesvirinae/physiology , Gene Deletion , Germinal Center/immunology , Germinal Center/virology , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Tumor Virus Infections/genetics , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Virus Latency
5.
Genetics ; 219(4)2021 12 10.
Article in English | MEDLINE | ID: mdl-34747456

ABSTRACT

PRDM9 is a DNA-binding histone methyltransferase that designates and activates recombination hotspots in mammals by locally trimethylating lysines 4 and 36 of histone H3. In mice, we recently reported two independently produced point mutations at the same residue, Glu360Pro (Prdm9EP) and Glu360Lys (Prdm9EK), which severely reduce its H3K4 and H3K36 methyltransferase activities in vivo. Prdm9EP is slightly less hypomorphic than Prdm9EK, but both mutations reduce both the number and amplitude of PRDM9-dependent H3K4me3 and H3K36me3 peaks in spermatocytes. While both mutations cause infertility with complete meiotic arrest in males, Prdm9EP, but not Prdm9EK, is compatible with some female fertility. When we tested the effects of these mutations in vitro, both Prdm9EP and Prdm9EK abolished H3K4 and H3K36 methyltransferase activity in full-length PRDM9. However, in the isolated PRDM9 PR/SET domain, these mutations selectively compromised H3K36 methyltransferase activity, while leaving H3K4 methyltransferase activity intact. The difference in these effects on the PR/SET domain vs the full-length protein shows that PRDM9 is not an intrinsically modular enzyme; its catalytic domain is influenced by its tertiary structure and possibly by its interactions with DNA and other proteins in vivo. These two informative mutations illuminate the enzymatic chemistry of PRDM9, and potentially of PR/SET domains in general, reveal the minimal threshold of PRDM9-dependent catalytic activity for female fertility, and potentially have some practical utility for genetic mapping and genomics.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , PR-SET Domains , Animals , Catalytic Domain/genetics , Female , Fertility/genetics , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Infertility, Male/genetics , Male , Mice , Mutation , Structure-Activity Relationship
6.
Development ; 148(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34100066

ABSTRACT

Spermatogenesis is precisely controlled by complex gene-expression programs. During mammalian male germ-cell development, a crucial feature is the repression of transcription before spermatid elongation. Previously, we discovered that the RNA-binding protein EWSR1 plays an important role in meiotic recombination in mouse, and showed that EWSR1 is highly expressed in late meiotic cells and post-meiotic cells. Here, we used an Ewsr1 pachytene stage-specific knockout mouse model to study the roles of Ewsr1 in late meiotic prophase I and in spermatozoa maturation. We show that loss of EWSR1 in late meiotic prophase I does not affect proper meiosis completion, but does result in defective spermatid elongation and chromocenter formation in the developing germ cells. As a result, male mice lacking EWSR1 after pachynema are sterile. We found that, in Ewsr1 CKO round spermatids, transition from a meiotic gene-expression program to a post-meiotic and spermatid gene expression program related to DNA condensation is impaired, suggesting that EWSR1 plays an important role in regulation of spermiogenesis-related mRNA synthesis necessary for spermatid differentiation into mature sperm.


Subject(s)
RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Spermatids/metabolism , Spermatogenesis/genetics , Spermatogenesis/physiology , Animals , Gene Expression Regulation, Developmental , Male , Meiosis , Meiotic Prophase I , Mice , Mice, Knockout , Spermatozoa
8.
Mol Biol Cell ; 32(1): 1-14, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33175657

ABSTRACT

Meiotic recombination in most mammals requires recombination hotspot activation through the action of the histone 3 Lys-4 and Lys-36 methyltransferase PRDM9 to ensure successful double-strand-break initiation and repair. Here we show that EWSR1, a protein whose role in meiosis was not previously clarified in detail, binds to both PRDM9 and pREC8, a phosphorylated meiosis-specific cohesin, in male meiotic cells. We created a Ewsr1 conditional knockout mouse model to deplete EWSR1 before the onset of meiosis and found that absence of EWSR1 causes meiotic arrest with decreased histone trimethylation at meiotic hotspots, impaired DNA double-strand-break repair, and reduced crossover number. Our results demonstrate that EWSR1 is essential for promoting PRDM9-dependent histone methylation and normal meiotic progress, possibly by facilitating the linking between PRDM9-bound hotspots and the nascent chromosome axis through its component cohesin pREC8.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomes, Mammalian/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , RNA-Binding Protein EWS/metabolism , Recombination, Genetic/genetics , Animals , Chromosomal Proteins, Non-Histone , Crossing Over, Genetic , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Male , Meiosis , Methylation , Mice, Knockout , Protein Binding , Protein Multimerization , Spermatozoa/metabolism , Synaptonemal Complex/metabolism , Cohesins
9.
Entropy (Basel) ; 22(10)2020 Oct 21.
Article in English | MEDLINE | ID: mdl-33286955

ABSTRACT

We attempted to attain atomic-scale insights into the mechanism of the heat-induced phase transition of two thermoresponsive polymers containing amide groups, poly(N-isopropylacrylamide) (PNIPAM) and poly(2-isopropyl-2-oxazoline) (PIPOZ), and we succeeded in reproducing the existence of lower critical solution temperature (LCST). The simulation data are in accord with experimental findings. We found out that the entropy has an important contribution to the thermodynamics of the phase separation transition. Moreover, after decomposing further the entropy change to contributions from the solutes and from the solvent, it appeared out that the entropy of the solvent has the decisive share for the lowering of the free energy of the system when increasing the temperature above the LCST. Our conclusion is that the thermoresponsive behavior is driven by the entropy of the solvent. The water molecules structured around the functional groups of the polymer that are exposed to contact with the solvent in the extended conformation lower the enthalpy of the system, but at certain temperature the extended conformation of the polymer collapses as a result of dominating entropy gain from "released" water molecules. We stress also on the importance of using more than one reference molecule in the simulation box at the setup of the simulation.

10.
Sci Adv ; 6(43)2020 10.
Article in English | MEDLINE | ID: mdl-33097538

ABSTRACT

In many mammals, genomic sites for recombination are determined by the histone methyltransferase PRMD9. Some mouse strains lacking PRDM9 are infertile, but instances of fertility or semifertility in the absence of PRDM9 have been reported in mice, canines, and a human female. Such findings raise the question of how the loss of PRDM9 is circumvented to maintain fertility. We show that genetic background and sex-specific modifiers can obviate the requirement for PRDM9 in mice. Specifically, the meiotic DNA damage checkpoint protein CHK2 acts as a modifier allowing female-specific fertility in the absence of PRDM9. We also report that, in the absence of PRDM9, a PRDM9-independent recombination system is compatible with female meiosis and fertility, suggesting sex-specific regulation of meiotic recombination, a finding with implications for speciation.

11.
Genome Res ; 29(7): 1078-1086, 2019 07.
Article in English | MEDLINE | ID: mdl-31186301

ABSTRACT

A hallmark of meiosis is the rearrangement of parental alleles to ensure genetic diversity in the gametes. These chromosome rearrangements are mediated by the repair of programmed DNA double-strand breaks (DSBs) as genetic crossovers between parental homologs. In mice, humans, and many other mammals, meiotic DSBs occur primarily at hotspots, determined by sequence-specific binding of the PRDM9 protein. Without PRDM9, meiotic DSBs occur near gene promoters and other functional sites. Studies in a limited number of mouse strains showed that functional PRDM9 is required to complete meiosis, but despite its apparent importance, Prdm9 has been repeatedly lost across many animal lineages. Both the reason for mouse sterility in the absence of PRDM9 and the mechanism by which Prdm9 can be lost remain unclear. Here, we explore whether mice can tolerate the loss of Prdm9 By generating Prdm9 functional knockouts in an array of genetic backgrounds, we observe a wide range of fertility phenotypes and ultimately demonstrate that PRDM9 is not required for completion of male meiosis. Although DSBs still form at a common subset of functional sites in all mice lacking PRDM9, meiotic outcomes differ substantially. We speculate that DSBs at functional sites are difficult to repair as a crossover and that by increasing the efficiency of crossover formation at these sites, genetic modifiers of recombination rates can allow for meiotic progression. This model implies that species with a sufficiently high recombination rate may lose Prdm9 yet remain fertile.


Subject(s)
Histone-Lysine N-Methyltransferase/physiology , Meiosis , Animals , Female , Fertility/genetics , Fertility/physiology , Histone-Lysine N-Methyltransferase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatogenesis/physiology , X Chromosome
12.
Curr Biol ; 29(6): 1002-1018.e7, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30853435

ABSTRACT

Meiotic recombination is required for correct segregation of chromosomes to gametes and to generate genetic diversity. In mice and humans, DNA double-strand breaks (DSBs) are initiated by SPO11 at recombination hotspots activated by PRDM9-catalyzed histone modifications on open chromatin. However, the DSB-initiating and repair proteins are associated with a linear proteinaceous scaffold called the chromosome axis, the core of which is composed of cohesin proteins. STAG3 is a stromalin subunit common to all meiosis-specific cohesin complexes. Mutations of meiotic cohesin proteins, especially STAG3, perturb both axis formation and recombination in the mouse, prompting determination of how the processes are mechanistically related. Protein interaction and genetic analyses revealed that PRDM9 interacts with STAG3 and REC8 in cooperative relationships that promote normal levels of meiotic DSBs at recombination hotspots in spermatocytes. The efficacy of the Prdm9-Stag3 genetic interaction in promoting DSB formation depends on PRDM9-mediated histone methyltransferase activity. Moreover, STAG3 deficiency has a major effect on DSB number even in the absence of PRDM9, showing that its role is not restricted to canonical PRDM9-activated hotspots. STAG3 and REC8 promote axis localization of the DSB-promoting proteins HORMAD1, IHO1, and MEI4, as well as SPO11 activity. These results establish that PRDM9 and axis-associated cohesin complexes together coordinate and facilitate meiotic recombination by recruiting key proteins for initiation of DSBs, thereby associating activated hotspots with DSB-initiating complexes on the axis.


Subject(s)
Cell Cycle Proteins/genetics , DNA Breaks, Double-Stranded , Histone-Lysine N-Methyltransferase/genetics , Meiosis , Animals , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Male , Mice , Spermatocytes
13.
Genetics ; 211(3): 831-845, 2019 03.
Article in English | MEDLINE | ID: mdl-30593494

ABSTRACT

The epigenetic landscape varies greatly among cell types. Although a variety of writers, readers, and erasers of epigenetic features are known, we have little information about the underlying regulatory systems controlling the establishment and maintenance of these features. Here, we have explored how natural genetic variation affects the epigenome in mice. Studying levels of H3K4me3, a histone modification at sites such as promoters, enhancers, and recombination hotspots, we found tissue-specific trans-regulation of H3K4me3 levels in four highly diverse cell types: male germ cells, embryonic stem cells, hepatocytes, and cardiomyocytes. To identify the genetic loci involved, we measured H3K4me3 levels in male germ cells in a mapping population of 59 BXD recombinant inbred lines. We found extensive trans-regulation of H3K4me3 peaks, including six major histone quantitative trait loci (QTL). These chromatin regulatory loci act dominantly to suppress H3K4me3, which at hotspots reduces the likelihood of subsequent DNA double-strand breaks. QTL locations do not correspond with genes encoding enzymes known to metabolize chromatin features. Instead their locations match clusters of zinc finger genes, making these possible candidates that explain the dominant suppression of H3K4me3. Collectively, these data describe an extensive, set of chromatin regulatory loci that control the epigenetic landscape.


Subject(s)
Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Hepatocytes/metabolism , Histone Code , Myocytes, Cardiac/metabolism , Spermatogonia/metabolism , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , Female , Male , Mice , Mice, Inbred C57BL , Mutation , Organ Specificity , Quantitative Trait Loci , Recombination, Genetic
14.
Math Biosci Eng ; 15(4): 1011-1032, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30380319

ABSTRACT

Human illness due to contamination of food by pathogenic strains of Escherichia coli is a serious public health concern and can cause significant economic losses in the food industry. Recent outbreaks of such illness sourced from ground beef production motivates the work in this paper. Most ground beef is produced in large facilities where many carcasses are butchered and various pieces of them are ground together in sequential batches. Assuming that the source of contamination is a single carcass and that downstream from the production facility ground beef from a particular batch has been identified as contaminated by E. coli, the probability that previous and subsequent batches are also contaminated is modelled. This model may help the beef industry to identify the likelihood of contamination in other batches and potentially save money by not needing to cook or recall unaffected batches of ground beef.


Subject(s)
Escherichia coli/isolation & purification , Escherichia coli/pathogenicity , Food Microbiology/statistics & numerical data , Red Meat/microbiology , Animals , Cattle , Colony Count, Microbial , Disease Outbreaks/statistics & numerical data , Escherichia coli Infections/epidemiology , Humans , Likelihood Functions , Mathematical Concepts , Meat-Packing Industry , Probability , Product Recalls and Withdrawals
15.
PLoS Genet ; 14(10): e1007657, 2018 10.
Article in English | MEDLINE | ID: mdl-30365547

ABSTRACT

In most mammals, including mice and humans, meiotic recombination is determined by the meiosis specific histone methytransferase PRDM9, which binds to specific DNA sequences and trimethylates histone 3 at lysine-4 and lysine-36 at the adjacent nucleosomes. These actions ensure successful DNA double strand break formation and repair that occur on the proteinaceous structure forming the chromosome axis. The process of hotspot association with the axis after their activation by PRDM9 is poorly understood. Previously, we and others have identified CXXC1, an ortholog of S. cerevisiae Spp1 in mammals, as a PRDM9 interactor. In yeast, Spp1 is a histone methyl reader that links H3K4me3 sites with the recombination machinery, promoting DSB formation. Here, we investigated whether CXXC1 has a similar function in mouse meiosis. We created two Cxxc1 conditional knockout mouse models to deplete CXXC1 generally in germ cells, and before the onset of meiosis. Surprisingly, male knockout mice were fertile, and the loss of CXXC1 in spermatocytes had no effect on PRDM9 hotspot trimethylation, double strand break formation or repair. Our results demonstrate that CXXC1 is not an essential link between PRDM9-activated recombination hotspot sites and DSB machinery and that the hotspot recognition pathway in mouse is independent of CXXC1.


Subject(s)
DNA Breaks, Double-Stranded , Histone-Lysine N-Methyltransferase/metabolism , Trans-Activators/genetics , Animals , DNA/metabolism , DNA Repair , Germ Cells/physiology , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/metabolism , Homologous Recombination , Male , Meiosis/genetics , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatocytes/metabolism , Spermatocytes/physiology , Trans-Activators/metabolism
16.
Trends Genet ; 34(4): 291-300, 2018 04.
Article in English | MEDLINE | ID: mdl-29366606

ABSTRACT

PRDM9 is a zinc finger protein that binds DNA at specific locations in the genome where it trimethylates histone H3 at lysines 4 and 36 at surrounding nucleosomes. During meiosis in many species, including humans and mice where PRDM9 has been most intensely studied, these actions determine the location of recombination hotspots, where genetic recombination occurs. In addition, PRDM9 facilitates the association of hotspots with the chromosome axis, the site of the programmed DNA double-strand breaks (DSBs) that give rise to genetic exchange between chromosomes. In the absence of PRDM9 DSBs are not properly repaired. Collectively, these actions determine patterns of genetic linkage and the possibilities for chromosome reorganization over successive generations.


Subject(s)
Genome , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Meiosis , Recombination, Genetic , Animals , DNA Breaks, Double-Stranded , Eukaryotic Cells/cytology , Eukaryotic Cells/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Methylation , Mice , Nucleosomes/enzymology , Nucleosomes/genetics , Protein Domains
17.
Indian J Cancer ; 54(1): 73-81, 2017.
Article in English | MEDLINE | ID: mdl-29199668

ABSTRACT

Optimal management and the role of surgery in multimodality treatment for N2 disease nonsmall cell lung cancer (NSCLC) are controversial. In this review, we focus on the possible role of pneumonectomy as a justified procedure in patients with persistent N2 disease following induction therapy. We have conducted an OVID PubMedbased search including manuscripts published in English for relevant studies. The interpretation of these trials highlights the lack of clarity and consistency in our management and leaves areas of controversy. There are no Level 1 data to support either performing or not performing pneumonectomy in this setting. The majority of the literature reviewed stresses the high risk of mortality and morbidity following pneumonectomy as a part of a trimodality approach to Stage IIIA/N2 NSCLC disease. However, selected highvolume institutions do follow this strategy with the level of risk seemingly justifying it for a highly selected group of patients, and this approach to Stage III/N2 NSCLC can be offered safely with acceptable mortality. Patient selection, response rate to induction therapy, and R0 resection are crucial for survival in experienced centers.


Subject(s)
Carcinoma, Non-Small-Cell Lung/surgery , Lung Neoplasms/surgery , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Chemotherapy, Adjuvant/methods , Humans , Induction Chemotherapy/methods , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Neoadjuvant Therapy/methods , Neoplasm Staging/methods , Pneumonectomy/methods
18.
R Soc Open Sci ; 4(9): 170777, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28989780

ABSTRACT

In 1665, Huygens observed that two identical pendulum clocks, weakly coupled through a heavy beam, soon synchronized with the same period and amplitude but with the two pendula swinging in opposite directions. This behaviour is now called anti-phase synchronization. This paper presents an analysis of the behaviour of a large class of coupled identical oscillators, including Huygens' clocks, using methods of equivariant bifurcation theory. The equivariant normal form for such systems is developed and the possible solutions are characterized. The transformation of the physical system parameters to the normal form parameters is given explicitly and applied to the physical values appropriate for Huygens' clocks, and to those of more recent studies. It is shown that Huygens' physical system could only exhibit anti-phase motion, explaining why Huygens observed exclusively this. By contrast, some more recent researchers have observed in-phase or other more complicated motion in their own experimental systems. Here, it is explained which physical characteristics of these systems allow for the existence of these other types of stable solutions. The present analysis not only accounts for these previously observed solutions in a unified framework, but also introduces behaviour not classified by other authors, such as a synchronized toroidal breather and a chaotic toroidal breather.

19.
Mol Biol Cell ; 28(3): 488-499, 2017 Feb 01.
Article in English | MEDLINE | ID: mdl-27932493

ABSTRACT

In mammals, meiotic recombination occurs at 1- to 2-kb genomic regions termed hotspots, whose positions and activities are determined by PRDM9, a DNA-binding histone methyltransferase. We show that the KRAB domain of PRDM9 forms complexes with additional proteins to allow hotspots to proceed into the next phase of recombination. By a combination of yeast-two hybrid assay, in vitro binding, and coimmunoprecipitation from mouse spermatocytes, we identified four proteins that directly interact with PRDM9's KRAB domain, namely CXXC1, EWSR1, EHMT2, and CDYL. These proteins are coexpressed in spermatocytes at the early stages of meiotic prophase I, the limited period when PRDM9 is expressed. We also detected association of PRDM9-bound complexes with the meiotic cohesin REC8 and the synaptonemal complex proteins SYCP3 and SYCP1. Our results suggest a model in which PRDM9-bound hotspot DNA is brought to the chromosomal axis by the action of these proteins, ensuring the proper chromatin and spatial environment for subsequent recombination events.


Subject(s)
Chromosomes/physiology , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Animals , Chromatin/metabolism , Chromosomes/genetics , DNA/metabolism , DNA Breaks, Double-Stranded , Genome , Histone-Lysine N-Methyltransferase/physiology , Homologous Recombination , Male , Meiosis/physiology , Mice , Protein Domains , Recombination, Genetic/physiology , Spermatocytes/metabolism
20.
PLoS Genet ; 12(6): e1006146, 2016 06.
Article in English | MEDLINE | ID: mdl-27362481

ABSTRACT

In many mammals, including humans and mice, the zinc finger histone methyltransferase PRDM9 performs the first step in meiotic recombination by specifying the locations of hotspots, the sites of genetic recombination. PRDM9 binds to DNA at hotspots through its zinc finger domain and activates recombination by trimethylating histone H3K4 on adjacent nucleosomes through its PR/SET domain. Recently, the isolated PR/SET domain of PRDM9 was shown capable of also trimethylating H3K36 in vitro, raising the question of whether this reaction occurs in vivo during meiosis, and if so, what its function might be. Here, we show that full-length PRDM9 does trimethylate H3K36 in vivo in mouse spermatocytes. Levels of H3K4me3 and H3K36me3 are highly correlated at hotspots, but mutually exclusive elsewhere. In vitro, we find that although PRDM9 trimethylates H3K36 much more slowly than it does H3K4, PRDM9 is capable of placing both marks on the same histone molecules. In accord with these results, we also show that PRDM9 can trimethylate both K4 and K36 on the same nucleosomes in vivo, but the ratio of K4me3/K36me3 is much higher for the pair of nucleosomes adjacent to the PRDM9 binding site compared to the next pair further away. Importantly, H3K4me3/H3K36me3-double-positive nucleosomes occur only in regions of recombination: hotspots and the pseudoautosomal (PAR) region of the sex chromosomes. These double-positive nucleosomes are dramatically reduced when PRDM9 is absent, showing that this signature is PRDM9-dependent at hotspots; the residual double-positive nucleosomes most likely come from the PRDM9-independent PAR. These results, together with the fact that PRDM9 is the only known mammalian histone methyltransferase with both H3K4 and H3K36 trimethylation activity, suggest that trimethylation of H3K36 plays an important role in the recombination process. Given the known requirement of H3K36me3 for double strand break repair by homologous recombination in somatic cells, we suggest that it may play the same role in meiosis.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Homologous Recombination/genetics , Meiosis/genetics , Recombination, Genetic/genetics , Animals , Binding Sites/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Histone Methyltransferases , Histones/genetics , Mice , Mice, Inbred C57BL , Nucleosomes/genetics , Zinc Fingers/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...